Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Markus Hansson is active.

Publication


Featured researches published by Markus Hansson.


Nature Communications | 2015

Variants in ELL2 influencing immunoglobulin levels associate with multiple myeloma

Bhairavi Swaminathan; Guðmar Thorleifsson; Magnus Jöud; Mina Ali; Ellinor Johnsson; Ram Ajore; Patrick Sulem; Britt-Marie Halvarsson; Guðmundur Eyjolfsson; Vilhelmína Haraldsdóttir; Christina M. Hultman; Erik Ingelsson; Sigurður Yngvi Kristinsson; Anna K. Kähler; Stig Lenhoff; Gisli Masson; Ulf-Henrik Mellqvist; Robert Månsson; Sven Nelander; Isleifur Olafsson; Olof Sigurðardottir; Hlif Steingrimsdottir; Annette Juul Vangsted; Ulla Vogel; Anders Waage; Hareth Nahi; Daniel F. Gudbjartsson; Thorunn Rafnar; Ingemar Turesson; Urban Gullberg

Multiple myeloma (MM) is characterized by an uninhibited, clonal growth of plasma cells. While first-degree relatives of patients with MM show an increased risk of MM, the genetic basis of inherited MM susceptibility is incompletely understood. Here we report a genome-wide association study in the Nordic region identifying a novel MM risk locus at ELL2 (rs56219066T; odds ratio (OR)=1.25; P=9.6 × 10−10). This gene encodes a stoichiometrically limiting component of the super-elongation complex that drives secretory-specific immunoglobulin mRNA production and transcriptional regulation in plasma cells. We find that the MM risk allele harbours a Thr298Ala missense variant in an ELL2 domain required for transcription elongation. Consistent with a hypomorphic effect, we find that the MM risk allele also associates with reduced levels of immunoglobulin A (IgA) and G (IgG) in healthy subjects (P=8.6 × 10−9 and P=6.4 × 10−3, respectively) and, potentially, with an increased risk of bacterial meningitis (OR=1.30; P=0.0024).


The New England Journal of Medicine | 2015

Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma.

Abstr Act; Henk M. Lokhorst; Torben Plesner; Hareth Nahi; Peter Gimsing; Markus Hansson; Ulrik V. Lassen; Jakub Krejcik; A Palumbo; Tahamtan Ahmadi; I. Khan; Jianping Wang; N. Losic; Steen Lisby; Linda Basse; Nikolai C. Brun

BACKGROUND Multiple myeloma cells uniformly overexpress CD38. We studied daratumumab, a CD38-targeting, human IgG1κ monoclonal antibody, in a phase 1-2 trial involving patients with relapsed myeloma or relapsed myeloma that was refractory to two or more prior lines of therapy. METHODS In part 1, the dose-escalation phase, we administered daratumumab at doses of 0.005 to 24 mg per kilogram of body weight. In part 2, the dose-expansion phase, 30 patients received 8 mg per kilogram of daratumumab and 42 received 16 mg per kilogram, administered once weekly (8 doses), twice monthly (8 doses), and monthly for up to 24 months. End points included safety, efficacy, and pharmacokinetics. RESULTS No maximum tolerated dose was identified in part 1. In part 2, the median time since diagnosis was 5.7 years. Patients had received a median of four prior treatments; 79% of the patients had disease that was refractory to the last therapy received (64% had disease refractory to proteasome inhibitors and immunomodulatory drugs and 64% had disease refractory to bortezomib and lenalidomide), and 76% had received autologous stem-cell transplants. Infusion-related reactions in part 2 were mild (71% of patients had an event of any grade, and 1% had an event of grade 3), with no dose-dependent adverse events. The most common adverse events of grade 3 or 4 (in ≥ 5% of patients) were pneumonia and thrombocytopenia. The overall response rate was 36% in the cohort that received 16 mg per kilogram (15 patients had a partial response or better, including 2 with a complete response and 2 with a very good partial response) and 10% in the cohort that received 8 mg per kilogram (3 had a partial response). In the cohort that received 16 mg per kilogram, the median progression-free survival was 5.6 months (95% confidence interval [CI], 4.2 to 8.1), and 65% (95% CI, 28 to 86) of the patients who had a response did not have progression at 12 months. CONCLUSIONS Daratumumab monotherapy had a favorable safety profile and encouraging efficacy in patients with heavily pretreated and refractory myeloma. (Funded by Janssen Research and Development and Genmab; ClinicalTrials.gov number, NCT00574288.).


The New England Journal of Medicine | 2016

Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma

Philippe Moreau; Tamas Masszi; Norbert Grzasko; Nizar J. Bahlis; Markus Hansson; Luděk Pour; Irwindeep Sandhu; Peter Ganly; Bartrum Baker; Sharon Jackson; Anne-Marie Stoppa; David R Simpson; Peter Gimsing; A Palumbo; L. Garderet; Michele Cavo; Shaji Kumar; Cyrille Touzeau; Francis Buadi; Jacob P. Laubach; Deborah Berg; Jianchang Lin; A. Di Bacco; Ai-Min Hui; H van de Velde; Paul G. Richardson; Eric Kupperman; Allison Berger; Larry Dick; Mark Williamson

BACKGROUND Ixazomib is an oral proteasome inhibitor that is currently being studied for the treatment of multiple myeloma. METHODS In this double-blind, placebo-controlled, phase 3 trial, we randomly assigned 722 patients who had relapsed, refractory, or relapsed and refractory multiple myeloma to receive ixazomib plus lenalidomide-dexamethasone (ixazomib group) or placebo plus lenalidomide-dexamethasone (placebo group). The primary end point was progression-free survival. RESULTS Progression-free survival was significantly longer in the ixazomib group than in the placebo group at a median follow-up of 14.7 months (median progression-free survival, 20.6 months vs. 14.7 months; hazard ratio for disease progression or death in the ixazomib group, 0.74; P=0.01); a benefit with respect to progression-free survival was observed with the ixazomib regimen, as compared with the placebo regimen, in all prespecified patient subgroups, including in patients with high-risk cytogenetic abnormalities. The overall rates of response were 78% in the ixazomib group and 72% in the placebo group, and the corresponding rates of complete response plus very good partial response were 48% and 39%. The median time to response was 1.1 months in the ixazomib group and 1.9 months in the placebo group, and the corresponding median duration of response was 20.5 months and 15.0 months. At a median follow-up of approximately 23 months, the median overall survival has not been reached in either study group, and follow-up is ongoing. The rates of serious adverse events were similar in the two study groups (47% in the ixazomib group and 49% in the placebo group), as were the rates of death during the study period (4% and 6%, respectively); adverse events of at least grade 3 severity occurred in 74% and 69% of the patients, respectively. Thrombocytopenia of grade 3 and grade 4 severity occurred more frequently in the ixazomib group (12% and 7% of the patients, respectively) than in the placebo group (5% and 4% of the patients, respectively). Rash occurred more frequently in the ixazomib group than in the placebo group (36% vs. 23% of the patients), as did gastrointestinal adverse events, which were predominantly low grade. The incidence of peripheral neuropathy was 27% in the ixazomib group and 22% in the placebo group (grade 3 events occurred in 2% of the patients in each study group). Patient-reported quality of life was similar in the two study groups. CONCLUSIONS The addition of ixazomib to a regimen of lenalidomide and dexamethasone was associated with significantly longer progression-free survival; the additional toxic effects with this all-oral regimen were limited. (Funded by Millennium Pharmaceuticals; TOURMALINE-MM1 ClinicalTrials.gov number, NCT01564537.).


Journal of Interferon and Cytokine Research | 1999

HISTAMINE PROTECTS T CELLS AND NATURAL KILLER CELLS AGAINST OXIDATIVE STRESS

Markus Hansson; Svante Hermodsson; Mats Brune; Ulf-Henrik Mellqvist; Peter Naredi; Åsa Betten; Kurt R. Gehlsen; Kristoffer Hellstrand

Oxidative stress inflicted by monocytes/macrophages (MO) is recognized as an important immunosuppressive mechanism in human neoplastic disease. We report that two types of lymphocytes of relevance for protection against malignant cells, T cells and natural killer (NK) cells, became anergic to the T cell and NK cell activator interleukin-2 (IL-2) after exposure to MO-derived reactive oxygen metabolites and subsequently acquired features characteristic of apoptosis. The MO-induced anergy and apoptosis in T cells and NK cells were reversed by histamine, an inhibitor of reactive oxygen metabolite synthesis in MO. We propose that strategies to circumvent oxidative inhibition of lymphocytes may be of benefit in immunotherapy of neoplastic disease.


European Journal of Haematology | 2009

NK cell‐mediated killing of AML blasts: role of histamine, monocytes and reactive oxygen metabolites

Mats Brune; Markus Hansson; Ulf-Henrik Mellqvist; S. Hermodsson; Kristoffer Hellstrand

Abstract:  Blasts recovered from patients with acute myelogenous leukaemia (AML) were lysed by heterologous natural killer (NK) cells treated with NK cell‐activating cytokines such as interleukin‐2 (IL‐2) or interferon‐α (IFN‐α). The cytokine‐induced killing of AML blasts was inhibited by monocytes, recovered from peripheral blood by counterflow centrifugal elutriation. Histamine, at concentrations exceeding 0.1 μM, abrogated the monocyte‐induced inhibition of NK cells; thereby, histamine and IL‐2 or histamine and IFN‐α synergistically induced NK cell‐mediated destruction of AML blasts. The effect of histamine was completely blocked by the histamine H2‐receptor (H2R) antagonist ranitidine but not by its chemical control AH20399AA. Catalase, a scavenger of reactive oxygen metabolites (ROM), reversed the monocyte‐induced inhibition of NK cell‐mediated killing of blast cells, indicating that the inhibitory signal was mediated by products of the respiratory burst of monocytes. It is concluded that (i) monocytes inhibit anti‐leukemic properties of NK cells, (ii) the inhibition is conveyed by monocyte‐derived ROM, and (iii) histamine reverses the inhibitory signal and, thereby, synergizes with NK cell‐activating cytokines to induce killing of AML blasts.


Leukemia | 2014

Expert panel consensus statement on the optimal use of pomalidomide in relapsed and refractory multiple myeloma

Meletios A. Dimopoulos; Xavier Leleu; Antonio Palumbo; P. Moreau; Michel Delforge; Michele Cavo; Heinz Ludwig; Gareth J. Morgan; Faith E. Davies; Pieter Sonneveld; Steve Schey; Sonja Zweegman; Markus Hansson; Katja Weisel; M.V. Mateos; Thierry Facon; Jesús F. San Miguel

In this report, a panel of European myeloma experts discuss the role of pomalidomide in the treatment of relapsed and refractory multiple myeloma (RRMM). Based on the available evidence, the combination of pomalidomide and low-dose dexamethasone is a well-tolerated and effective treatment option for patients with RRMM who have exhausted treatment with lenalidomide and bortezomib. The optimal starting dose of pomalidomide is 4 mg given on days 1–21 of each 28-day cycle, whereas dexamethasone is administered at a dose of 40 mg weekly (reduced to 20 mg for patients aged >75 years). The treatment should continue until evidence of disease progression or unacceptable toxicity. Dose-modification schemes have been established for patients who develop neutropenia, thrombocytopaenia and other grade 3–4 adverse events during pomalidomide therapy. Guidance on the prevention and management of infections and venous thromboembolism is provided, based on the available clinical evidence and the experience of panel members. The use of pomalidomide in special populations, such as patients with advanced age, renal impairment or unfavourable cytogenetic features, is also discussed.


Cancer Investigation | 2000

Histamine: A Novel Approach to Cancer Immunotherapy

Kristoffer Hellstrand; Mats Brune; Peter Naredi; Ulf-Henrik Mellqvist; Markus Hansson; Kurt R. Gehlsen; Svante Hermodsson

Abstract The functions of intratumoral lymphocytes in many human malignant tumors are inhibited by reactive oxygen species (ROS), generated by adjacent monocytes/macrophages (MO). In vitro data suggest that immunotherapeutic cytokines such as interleukin-2 (IL-2) or interferon-α (IFN-α) only weakly activate T cells or natural killer (NK) cells in a reconstituted environment of oxidative stress and that inhibitors of the formation of ROS or scavengers of ROS synergize with IL-2 and IFN-α to activate T cells and NK cells. In this review, we focus on the immunoenhancing properties ofhistamine, a biogenic amine. Histamine inhibits ROS formation in MO via H2–receptors; thereby, histamine protects NK cells from MO-mediated inhibition and synergizes with IL-2 and IFN-α to induce killing of NK cell-sensitive human tumor cells in vitro. Histamine also optimizes cytokine-induced activation of several subsets of T cells by affording protection against MO-inflicted oxidative inhibition. The putative clinical benefit of histamine as an adjunct to immunotherapy with IL-2 and/or IFN-α is currently evaluated in clinical trials in metastatic malignant melanoma and acute myelogenous leukemia.


Blood | 2016

Safety and efficacy of pomalidomide plus low-dose dexamethasone in STRATUS (MM-010): a phase 3b study in refractory multiple myeloma.

Meletios A. Dimopoulos; Antonio Palumbo; Paolo Corradini; Michele Cavo; Michel Delforge; Francesco Di Raimondo; Katja Weisel; Albert Oriol; Markus Hansson; Angelo Vacca; María Jesús Blanchard; Hartmut Goldschmidt; Chantal Doyen; Martin Kaiser; Mario Petrini; Pekka Anttila; Anna Maria Cafro; Reinier Raymakers; Jesús F. San-Miguel; Felipe de Arriba; Stefan Knop; Christoph Röllig; Enrique M. Ocio; Gareth J. Morgan; Neil Miller; Mathew Simcock; Teresa Peluso; Jennifer Herring; Lars Sternas; Mohamed H. Zaki

Patients with relapsed and/or refractory multiple myeloma (RRMM) have poor prognosis. The STRATUS study assessed safety and efficacy of pomalidomide plus low-dose dexamethasone in the largest cohort to date of patients with RRMM. Patients who failed treatment with bortezomib and lenalidomide and had adequate prior alkylator therapy were eligible. Pomalidomide 4 mg was given on days 1-21 of 28-day cycles with low-dose dexamethasone 40 mg (20 mg for patients aged >75 years) on days 1, 8, 15, and 22 until progressive disease or unacceptable toxicity. Safety was the primary end point; secondary end points included overall response rate (ORR), duration of response (DOR), progression-free survival (PFS), and overall survival (OS). Among 682 patients enrolled, median age was 66 years, and median time since diagnosis was 5.3 years. Median number of prior regimens was 5. Most patients were refractory to both lenalidomide and bortezomib (80.2%). Median follow-up was 16.8 months; median duration of treatment was 4.9 months. Most frequent grade 3/4 treatment-emergent adverse events were hematologic (neutropenia [49.7%], anemia [33.0%], and thrombocytopenia [24.1%]). Most common grade 3/4 nonhematologic toxicities were pneumonia (10.9%) and fatigue (5.9%). Grade 3/4 venous thromboembolism and peripheral neuropathy were rare (1.6% each). The ORR was 32.6%, and the median DOR was 7.4 months. Median PFS and OS were 4.6 months and 11.9 months, respectively. We present the largest trial to date evaluating pomalidomide plus low-dose dexamethasone in patients with RRMM, further confirming that this regimen offers clinically meaningful benefit and is generally well tolerated. www.Clinicaltrials.gov identifier NCT01712789.


Haematologica | 2012

Remission maintenance in acute myeloid leukemia: impact of functional histamine H2 receptors expressed by leukemic cells

Johan Aurelius; Anna Martner; Mats Brune; Lars Palmqvist; Markus Hansson; Kristoffer Hellstrand; Fredrik B. Thorén

Post-consolidation immunotherapy with histamine dihydrochloride and interleukin-2 has been shown to improve leukemia-free survival in acute myeloid leukemia in a phase III trial. For this study, treatment efficacy was determined among 145 trial patients with morphological forms of acute myeloid leukemia as defined by the French-American-British classification. Leukemia-free survival was strongly improved in M4/M5 (myelomonocytic/monocytic) leukemia but not in M2 (myeloblastic) leukemia. We also analyzed histamine H2 receptor expression by leukemic cells recovered from 26 newly diagnosed patients. H2 receptors were typically absent from M2 cells but frequently expressed by M4/M5 cells. M4/M5 cells, but not M2 cells, produced reactive oxygen species that triggered apoptosis in adjacent natural killer cells. These events were significantly inhibited by histamine dihydrochloride. Our data demonstrate the presence of functional histamine H2 receptors on human AML cells and suggest that expression of these receptors by leukemic cells may impact on the effectiveness of histamine-based immunotherapy.


Blood | 2012

Monocytic AML cells inactivate antileukemic lymphocytes: role of NADPH oxidase/gp91(phox) expression and the PARP-1/PAR pathway of apoptosis.

Johan Aurelius; Fredrik B. Thorén; Ali A. Akhiani; Mats Brune; Lars Palmqvist; Markus Hansson; Kristoffer Hellstrand; Anna Martner

Dysfunction of T cells and natural killer (NK) cells has been proposed to determine the course of disease in acute myeloid leukemia (AML), but only limited information is available on the mechanisms of lymphocyte inhibition. We aimed to evaluate to what extent human malignant AML cells use NADPH oxidase-derived reactive oxygen species (ROS) as an immune evasion strategy. We report that a subset of malignant myelomonocytic and monocytic AML cells (French-American-British [FAB] classes M4 and M5, respectively), recovered from blood or BM of untreated AML patients at diagnosis, expressed the NADPH oxidase component gp91(phox). Highly purified FAB M4/M5 AML cells produced large amounts of ROS on activation and triggered poly-[ADP-ribose] polymerase-1-dependent apoptosis in adjacent NK cells, CD4(+) T cells, and CD8(+) T cells. In contrast, immature (FAB class M1) and myeloblastic (FAB class M2) AML cells rarely expressed gp91(phox), did not produce ROS, and did not trigger NK or T-cell apoptosis. Microarray data from 207 AML patients confirmed a greater expression of gp91(phox) mRNA by FAB-M4/M5 AML cells than FAB-M1 cells (P < 10(-11)) or FAB-M2 cells (P < 10(-9)). Our data are suggestive of a novel mechanism by which monocytic AML cells evade cell-mediated immunity.

Collaboration


Dive into the Markus Hansson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ulf-Henrik Mellqvist

Sahlgrenska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Gimsing

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michel Delforge

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Katja Weisel

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar

Anders Waage

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Meletios A. Dimopoulos

National and Kapodistrian University of Athens

View shared research outputs
Researchain Logo
Decentralizing Knowledge